Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Metronomic chemotherapy plus anti-PD-1 in metastatic breast cancer: a Bayesian adaptive randomized phase 2 trial

Abstract

It remains unclear whether metronomic chemotherapy is superior to conventional chemotherapy when combined with immune checkpoint blockade. Here we performed a phase 2 clinical trial of metronomic chemotherapy combined with PD-1 blockade to compare the efficacy of combined conventional chemotherapy and PD-1 blockade using Bayesian adaptive randomization and efficacy monitoring. Eligible patients had metastatic HER2-negative breast cancer and had not received more than one prior line of standard chemotherapy. Patients (total n = 97) were randomized to receive (1) metronomic vinorelbine (NVB) monotherapy (n = 11), (2) NVB plus anti-PD-1 toripalimab (n = 7), (3) anti-angiogenic bevacizumab, NVB and toripalimab (n = 27), (4) conventional cisplatin, NVB and toripalimab (n = 26), or (5) metronomic cyclophosphamide, capecitabine, NVB and toripalimab (the VEX cohort) (n = 26). The primary endpoint was disease control rate (DCR). Secondary objectives included progression-free survival (PFS) and safety. The study met the primary endpoint. The VEX (69.7%) and cisplatin (73.7%) cohorts had the highest DCR. The median PFS of patients in the VEX cohort was the longest, reaching 6.6 months, followed by the bevacizumab (4.0 months) and cisplatin (3.5 months) cohorts. In general, the five regimens were well tolerated, with nausea and neutropenia being the most common adverse events. An exploratory mass cytometry analysis indicated that metronomic VEX chemotherapy reprograms the systemic immune response. Together, the clinical and translational data of this study indicate that metronomic VEX chemotherapy combined with PD-1 blockade can be a treatment option in patients with breast cancer. ClinicalTrials.gov Identifier: NCT04389073.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Trial design.
Fig. 2: Anti-tumor activity after treatment in the five cohorts.
Fig. 3: Subgroup analysis according to the clinical characteristics of the patients.
Fig. 4: Metronomic VEX chemotherapy specifically reprograms systemic immune characteristics.

Similar content being viewed by others

Data availability

Complete de-identified patient data will be available indefinitely within 2 years of the patients’ last survival follow-up visit and will be uploaded to ClinicalTrials.gov. Single-cell sequencing data can be obtained from the Gene Expression Omnibus with accession number GSE169246. The data of human reference genome 19 can be downloaded from UCSC Xena. Any additional information required to reanalyze the data reported in this paper is available from the corresponding author upon request, with detailed proposals for the use of the information. In response to the inquiry, please be informed that the timeframe for responding to requests is approximately 2 weeks.

References

  1. Hegde, P. S. & Chen, D. S. Top 10 challenges in cancer immunotherapy. Immunity 52, 17–35 (2020).

    Article  CAS  PubMed  Google Scholar 

  2. Mo, H. et al. Safety, anti-tumour activity, and pharmacokinetics of fixed-dose SHR-1210, an anti-PD-1 antibody in advanced solid tumours: a dose-escalation, phase 1 study. Br. J. Cancer 119, 538–545 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Carrick, S. et al. Single agent versus combination chemotherapy for metastatic breast cancer. Cochrane Database Syst. Rev. 2009, CD003372 (2009).

    PubMed  PubMed Central  Google Scholar 

  4. Moy, B. et al. Chemotherapy and targeted therapy for patients with human epidermal growth factor receptor 2-negative metastatic breast cancer that is either endocrine-pretreated or hormone receptor-negative: ASCO guideline update. J. Clin. Oncol. 39, 3938–3958 (2021).

    Article  CAS  PubMed  Google Scholar 

  5. Ozaki, Y. et al. Safety and efficacy of nivolumab plus bevacizumab, paclitaxel for HER2-negative metastatic breast cancer: primary results and biomarker data from a phase 2 trial (WJOG9917B). Eur. J. Cancer 171, 193–202 (2022).

    Article  CAS  PubMed  Google Scholar 

  6. Cortes, J. et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet 396, 1817–1828 (2020).

    Article  PubMed  Google Scholar 

  7. Jiang, Z. et al. Toripalimab plus nab-paclitaxel in metastatic or recurrent triple-negative breast cancer: a randomized phase 3 trial. Nat. Med. 30, 249–256 (2024).

    Article  CAS  PubMed  Google Scholar 

  8. Zhang, Y. et al. Single-cell analyses reveal key immune cell subsets associated with response to PD-L1 blockade in triple-negative breast cancer. Cancer Cell 39, 1578–1593 (2021).

    Article  CAS  PubMed  Google Scholar 

  9. Miles, D. W. et al. First-line bevacizumab in combination with chemotherapy for HER2-negative metastatic breast cancer: pooled and subgroup analyses of data from 2447 patients. Ann. Oncol. 24, 2773–2780 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Pasquier, E., Kavallaris, M. & André, N. Metronomic chemotherapy: new rationale for new directions. Nat. Rev. Clin. Oncol. 7, 455–465 (2010).

    Article  PubMed  Google Scholar 

  11. André, N., Carré, M. & Pasquier, E. Metronomics: towards personalized chemotherapy? Nat. Rev. Clin. Oncol. 11, 413–431 (2014).

    Article  PubMed  Google Scholar 

  12. Orecchioni, S. et al. Vinorelbine, cyclophosphamide and 5-FU effects on the circulating and intratumoural landscape of immune cells improve anti-PD-L1 efficacy in preclinical models of breast cancer and lymphoma. Br. J. Cancer 118, 1329–1336 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Munzone, E. et al. Efficacy of metronomic oral vinorelbine, cyclophosphamide, and capecitabine vs weekly intravenous paclitaxel in patients with estrogen receptor-positive, ERBB2-negative metastatic breast cancer: final results from the phase 2 METEORA-II randomized clinical trial. JAMA Oncol. 9, 1267–1272 (2023).

    Article  PubMed  Google Scholar 

  14. Gubin, M. M. et al. High-dimensional analysis delineates myeloid and lymphoid compartment remodeling during successful immune-checkpoint cancer therapy. Cell 175, 1014–1030 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Onkar, S. S. et al. The great immune escape: understanding the divergent immune response in breast cancer subtypes. Cancer Discov. 13, 23–40 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Shaked, Y. The pro-tumorigenic host response to cancer therapies. Nat. Rev. Cancer 19, 667–685 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Ho, W. J. et al. Integrated immunological analysis of a successful conversion of locally advanced hepatocellular carcinoma to resectability with neoadjuvant therapy. J. Immunother. Cancer 8, e000932 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Hiam-Galvez, K. J., Allen, B. M. & Spitzer, M. H. Systemic immunity in cancer. Nat. Rev. Cancer 21, 345–359 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Monjazeb, A. M. et al. A randomized trial of combined PD-L1 and CTLA-4 inhibition with targeted low-dose or hypofractionated radiation for patients with metastatic colorectal cancer. Clin. Cancer Res. 27, 2470–2480 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Simonds, E. F. et al. Deep immune profiling reveals targetable mechanisms of immune evasion in immune checkpoint inhibitor-refractory glioblastoma. J. Immunother. Cancer 9, e002181 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Lheureux, S. et al. Translational randomized phase II trial of cabozantinib in combination with nivolumab in advanced, recurrent, or metastatic endometrial cancer. J. Immunother. Cancer 10, e004233 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Rochigneux, P. et al. Mass cytometry reveals classical monocytes, NK cells, and ICOS+ CD4+ T cells associated with pembrolizumab efficacy in patients with lung cancer. Clin. Cancer Res. 28, 5136–5148 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chapman, A. et al. CD11b+CD28CD4+ human T cells: activation requirements and association with HLA-DR alleles. J. Immunol. 157, 4771–4780 (1996).

    Article  CAS  PubMed  Google Scholar 

  24. Markovic-Plese, S., Cortese, I., Wandinger, K. P., McFarland, H. F. & Martin, R. CD4+CD28 costimulation-independent T cells in multiple sclerosis. J. Clin. Investig. 108, 1185–1194 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Alegre, M.-L., Frauwirth, K. A. & Thompson, C. B. T-cell regulation by CD28 and CTLA-4. Nat. Rev. Immunol. 1, 220–228 (2001).

    Article  CAS  PubMed  Google Scholar 

  26. Wendel, M., Galani, I. E., Suri-Payer, E. & Cerwenka, A. Natural killer cell accumulation in tumors is dependent on IFN-γ and CXCR3 ligands. Cancer Res. 68, 8437–8445 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Wu, M., Fang, H. & Hwang, S. T. Cutting edge: CCR4 mediates antigen-primed T cell binding to activated dendritic cells. J. Immunol. 167, 4791–4795 (2001).

    Article  CAS  PubMed  Google Scholar 

  28. Bromley, S. K., Thomas, S. Y. & Luster, A. D. Chemokine receptor CCR7 guides T cell exit from peripheral tissues and entry into afferent lymphatics. Nat. Immunol. 6, 895–901 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Gregory, J. L. et al. Macrophage migration inhibitory factor induces macrophage recruitment via CC chemokine ligand 2. J. Immunol. 177, 8072–8079 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. Yang, R. et al. Galectin-9 interacts with PD-1 and TIM-3 to regulate T cell death and is a target for cancer immunotherapy. Nat. Commun. 12, 832 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Sugimoto, M. A., Vago, J. P., Teixeira, M. M. & Sousa, L. P. Annexin A1 and the resolution of inflammation: modulation of neutrophil recruitment, apoptosis, and clearance. J. Immunol. Res. 2016, 8239258 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Socinski, M. A. et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N. Engl. J. Med. 378, 2288–2301 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Shitara, K. et al. Efficacy and safety of pembrolizumab or pembrolizumab plus chemotherapy vs chemotherapy alone for patients with first-line, advanced gastric cancer: the KEYNOTE-062 phase 3 randomized clinical trial. JAMA Oncol. 6, 1571–1580 (2020).

    Article  PubMed  Google Scholar 

  34. Song, Y. et al. First-line serplulimab or placebo plus chemotherapy in PD-L1-positive esophageal squamous cell carcinoma: a randomized, double-blind phase 3 trial. Nat. Med. 29, 473–482 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Colombo, N. et al. Pembrolizumab for persistent, recurrent, or metastatic cervical cancer. N. Engl. J. Med. 385, 1856–1867 (2021).

    Article  CAS  PubMed  Google Scholar 

  36. Zhang, Z. et al. A real-world study of immune checkpoint inhibitors in advanced triple-negative breast cancer. Cancer Innov. 2, 172–180 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Miles, D. et al. Primary results from IMpassion131, a double-blind, placebo-controlled, randomised phase III trial of first-line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple-negative breast cancer. Ann. Oncol. 32, 994–1004 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. Schmid, P. et al. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 21, 44–59 (2020).

    Article  CAS  PubMed  Google Scholar 

  39. Schmid, P. et al. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N. Engl. J. Med. 379, 2108–2121 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Vicier, C. et al. MOVIE: a phase I, open-label, multicenter study to evaluate the safety and tolerability of metronomic vinorelbine combined with durvalumab plus tremelimumab in patients with advanced solid tumors. ESMO Open 7, 100646 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Patil, V. M. et al. Low-dose immunotherapy in head and neck cancer: a randomized study. J. Clin. Oncol. 41, 222–232 (2023).

    Article  CAS  PubMed  Google Scholar 

  42. Zsiros, E. et al. Efficacy and safety of pembrolizumab in combination with bevacizumab and oral metronomic cyclophosphamide in the treatment of recurrent ovarian cancer: a phase 2 nonrandomized clinical trial. JAMA Oncol. 7, 78–85 (2021).

    Article  PubMed  Google Scholar 

  43. Vergnenegre, A. et al. Safety and efficacy of second-line metronomic oral vinorelbine-atezolizumab combination in stage IV non-small-cell lung cancer: an open-label phase II trial (VinMetAtezo). Lung Cancer 178, 191–197 (2023).

    Article  CAS  PubMed  Google Scholar 

  44. Cortes, J. et al. Pembrolizumab plus chemotherapy in advanced triple-negative breast cancer. N. Engl. J. Med. 387, 217–226 (2022).

    Article  CAS  PubMed  Google Scholar 

  45. André, N. et al. Mathematical model of cancer growth controled by metronomic chemotherapies. ESAIM Proc. 41, 77–94 (2013).

    Article  Google Scholar 

  46. Huang, Y. et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc. Natl Acad. Sci. USA 109, 17561–17566 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Curigliano, G. et al. Antiangiogenic therapy in recurrent breast cancer with lymphangitic spread to the chest wall: a randomized phase II trial of bevacizumab with sequential or concurrent oral vinorelbine and capecitabine. Breast 24, 263–271 (2015).

    Article  PubMed  Google Scholar 

  48. Xu, B. et al. Dalpiciclib or placebo plus fulvestrant in hormone receptor-positive and HER2-negative advanced breast cancer: a randomized, phase 3 trial. Nat. Med. 27, 1904–1909 (2021).

    Article  CAS  PubMed  Google Scholar 

  49. Muss, H. B. et al. Toxicity of older and younger patients treated with adjuvant chemotherapy for node-positive breast cancer: the Cancer and Leukemia Group B Experience. J. Clin. Oncol. 25, 3699–3704 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Montagna, E. et al. Safety and efficacy study of metronomic vinorelbine, cyclophosphamide plus capecitabine in metastatic breast cancer: a phase II trial. Cancer Lett. 400, 276–281 (2017).

    Article  CAS  PubMed  Google Scholar 

  51. Butler, C. C. et al. Molnupiravir plus usual care versus usual care alone as early treatment for adults with COVID-19 at increased risk of adverse outcomes (PANORAMIC): an open-label, platform-adaptive randomised controlled trial. Lancet 401, 281–293 (2023).

    Article  CAS  PubMed  Google Scholar 

  52. Reis, G. et al. Effect of early treatment with ivermectin among patients with Covid-19. N. Engl. J. Med. 386, 1721–1731 (2022).

    Article  CAS  PubMed  Google Scholar 

  53. Puduvalli, V. K. et al. A Bayesian adaptive randomized phase II multicenter trial of bevacizumab with or without vorinostat in adults with recurrent glioblastoma. Neuro Oncol. 22, 1505–1515 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Voorwerk, L. et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial. Nat. Med. 25, 920–928 (2019).

    Article  CAS  PubMed  Google Scholar 

  55. Debien, V. et al. Immunotherapy in breast cancer: an overview of current strategies and perspectives. npj Breast Cancer 9, 7 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Kirchhammer, N., Trefny, M. P., Auf der Maur, P., Läubli, H. & Zippelius, A. Combination cancer immunotherapies: emerging treatment strategies adapted to the tumor microenvironment. Sci. Transl. Med. 14, eabo3605 (2022).

    Article  CAS  PubMed  Google Scholar 

  57. Galon, J. & Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 18, 197–218 (2019).

    Article  CAS  PubMed  Google Scholar 

  58. Rahim, M. K. et al. Dynamic CD8+ T cell responses to cancer immunotherapy in human regional lymph nodes are disrupted in metastatic lymph nodes. Cell 186, 1127–1143 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Peng, H. et al. Neoadjuvant FOLFIRINOX therapy is associated with increased effector T cells and reduced suppressor cells in patients with pancreatic cancer. Clin. Cancer Res. 27, 6761–6771 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Chong, E. A. et al. Pembrolizumab for B-cell lymphomas relapsing after or refractory to CD19-directed CAR T-cell therapy. Blood 139, 1026–1038 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Berry, D. A. Adaptive clinical trials in oncology. Nat. Rev. Clin. Oncol. 9, 199–207 (2011).

    Article  PubMed  Google Scholar 

  62. Berry, D. A. Bayesian clinical trials. Nat. Rev. Drug Discov. 5, 27–36 (2006).

    Article  CAS  PubMed  Google Scholar 

  63. Yu, S. et al. Distinct immune signatures discriminate between asymptomatic and presymptomatic SARS-CoV-2pos subjects. Cell Res. 31, 1148–1162 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Zhang, Q. et al. Mass cytometry-based peripheral blood analysis as a novel tool for early detection of solid tumours: a multicentre study. Gut 72, 996–1006 (2023).

    Article  CAS  PubMed  Google Scholar 

  65. Zunder, E. R. et al. Palladium-based mass tag cell barcoding with a doublet-filtering scheme and single-cell deconvolution algorithm. Nat. Protoc. 10, 316–333 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Finck, R. et al. Normalization of mass cytometry data with bead standards. Cytometry A 83, 483–494 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Samusik, N., Good, Z., Spitzer, M. H., Davis, K. L. & Nolan, G. P. Automated mapping of phenotype space with single-cell data. Nat. Methods 13, 493–496 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Berry, S. M., Carlin, B. P., Lee, J. J. & Muller, P. Bayesian Adaptive Methods for Clinical Trials (CRC Press, 2010).

  69. Harari, O. et al. Utilizing Bayesian predictive power in clinical trial design. Pharm. Stat. https://doi.org/10.1002/pst.2073 (2020).

Download references

Acknowledgements

We thank the patients, their families and the personnel involved in this trial. We thank Y. Yuan for his assistance with statistical analysis. This work was supported by the CAMS Innovation Fund for Medical Sciences (CIFMS 2021-I2M-1-014 to B.X. and CIFMS 2022-I2M-C&T-B-067 to H.M.), the National Natural Science Foundation of China (82230058 to F.M.) and the Beijing Hope Run Special Fund of Cancer Foundation of China (LC2021A08 to H.M.).

Author information

Authors and Affiliations

Authors

Contributions

H.M., Y.Y., H.G., L.Y., Y.W. and J.Z. participated in the design, execution and/or interpretation of the reported experiments or results. H.M., Y.Y., H.G., X.S., A.Z., J.W., X.G., L.Y., J.Z. and Y.W. participated in the sample acquisition or data analysis. H.M., Y.Y. and H.G. drafted the paper, with all authors contributing to the writing and providing feedback. F.M., B.X., X.Y. and H.Q. supervised all aspects of the research.

Corresponding authors

Correspondence to Haili Qian, Binghe Xu or Fei Ma.

Ethics declarations

Competing interests

X.B. reports personal advisory fees from Novartis and personal payment or honoraria fees for lecture from AstraZeneca, Pfizer, Roche and Eisai, outside of the submitted work. The other authors declare no competing interests.

Peer review

Peer review information

Nature Medicine thanks Aditya Bardia, Paolo Tarantino and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Saheli Sadanand, in collaboration with the Nature Medicine team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Clinical efficacy in patients receiving metronomic vinorelbine with or without PD-1 blockade.

Histograms showing the final probability distributions for the disease control rate (DCR) (a) and objective response rate (ORR) (b) as well as the Kaplan–Meier estimates of progression-free survival (PFS) (c) and overall survival (OS) (d). a, Among the patients receiving PD-1 blockade therapy, 54 achieved a controlled disease (DCR of 62.9%; 95% CI 52.5–72.7%), which compared favorably with the controlled DCR of 35.5% (95% CI 10.6–63.2%) in the NVB monotherapy cohort. b, ORR was 11.3% (95% CI 5.4–18.5%) in patients receiving PD-1 blockade therapy and 0% in the controlled NVB monotherapy cohort. c, Median PFS was 4.2 months (95% CI 3.2–5.3) in patients receiving PD-1 blockade therapy. In patients receiving metronomic chemotherapy with NVB alone, the median PFS was 1.4 months (95% CI 0.69–2.4). d, Median OS was 29.2 months (95% CI 19.9–41.7) in patients who received PD-1 blockade and 19.1 months (95% CI 6.6-47.2) in the control group.

Extended Data Fig. 2 Bayesian estimates of the disease control rate (DCR) according to the molecular subtype of patient, number of previous treatment lines, and presence of visceral metastasis.

Data are presented as the Bayesian estimates of DCR and 95% credible interval. TNBC, triple-negative breast cancer.

Extended Data Fig. 3 Specific reprogramming of systemic immune components is key to the efficacy of immunotherapy.

a, The t-distributed stochastic neighbor embedding (t-SNE) plots of immune cells sampled from patients receiving PD-1 blockade therapy, colored by major immune cell subsets. b, Heatmap displaying the median antigen intensity of markers used to generate a. c, Histograms of samples from baseline. None of the 32 CD45+ cell subsets was considerably different between responders (Pre-R) and nonresponders (Pre-NR). d, Principal component analysis (PCA) of samples before (Pre)and after treatment (Post). The combination therapy caused dramatic changes in the systemic immune profiles of patients. e, Comparison of the percentages of 32 identified cell subsets across the response group. f, Differential CTLA4 expression on T-cell subsets between responders (R) and nonresponders (NR). g, Differential expression of CD206 and CD33 on monocytes across the response group. In e and g, n=37 in Pre-R, n=20 in Pre-NR, n=35 in Post-R, n=22 in Post-NR. In e and g, exact P values by t- test (two-sided): *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. In e and g, boxes indicate median ± interquartile range; whiskers show minima and maxima.

Extended Data Fig. 4 Differential expression of cell markers according to the response groups.

a, Clusters 05, 30, and 31 were mainly from peripheral blood mononuclear cells (PBMCs) after treatment, whereas Clusters 03, 21, and 32 were primarily from baseline samples. b, RNA sequencing of baseline tumor tissues revealed that the infiltration of 28 immune cells in the tumor microenvironment did not correlate with immunotherapy efficacy (n=24 in R, n=14 in NR). c, PD-L1 expression (n=7 in R, n=5 in NR) and tumor mutation burden (n=12 in R, n=7 in NR) from baseline tumor samples showed no important difference between responders (R) and nonresponders (NR). Exact P values by two-sided Mann–Whitney test. d, Single-cell RNA sequencing of PBMCs from baseline samples did not show correlation between the subsets of PBMCs and the efficacy of immunotherapy (n=4 in R, n=7 in NR). e, Differences in subpopulation proportions and protein expression of T-cell subset clusters 03 and 05 between responders (R) and nonresponders (NR). f, Differences in subpopulation proportions and protein expression of monocytes Cluster 29 and Cluster 30 between responders (R) and nonresponders (NR). g, Differential expression of NK cell markers according to the response groups. In e-g, n=37 in Pre-R, n=20 in Pre-NR, n=35 in Post-R, n=22 in Post-NR. In e and f, representative histograms showing differentially expressed markers on cell subsets. In e, f and g, exact P values by t- test (two-sided): *P < 0.05, **P < 0.01. In b-d and f-g, boxes indicate median ± interquartile range; whiskers show minima and maxima.

Extended Data Fig. 5 Differential expression of cell markers according to the treatment cohorts.

a, Cells were consistently distributed across all major immune subsets at baseline. b, A consistently increase in the proportion of Cluster 04 and Cluster 14 was observed in all three cohorts. c, Differential expression of CCR7 on T-cell subsets in different treatment cohorts. d, Differential expression of the monocyte markers CD206 and CD14 after different treatments. B cell (e) and dendritic cell subsets (f) in patients did not change substantially after treatment. In b-f, n=17 in Pre-DDP, n=19 in Post-DDP; n=15 in Pre-BEV, n=16 in Post- BEV; n=25 in Pre-VEX, n=22 in Post-VEX. In b - d, exact P values by t- test (two-sided): *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. In b-f, boxes indicate median ± interquartile range; whiskers show minima and maxima.

Extended Data Fig. 6 Cell–cell communication between the peripheral blood mononuclear cell (PBMC) subsets.

a, Circos plots showing the multiple correlation matrix between the PBMC subset frequencies in CyTOF. b, Single-cell RNA sequencing analysis verified the negative correlation between monocytes and T-cell subsets. c, Analysis of cell–cell communication between T-cell subsets and monocytes. d, Analysis of cell–cell communication between monocytes and T-cell subsets. e, Difference of cell–cell communication between responders and nonresponders. In c-e, dot color reflects communication probabilities and dot size represents computed P values (one-sided permutation test) for interactions.

Extended Data Table 1 Probability of superiority among the five cohorts in all patients
Extended Data Table 2 Probability of superiority in the PD-1 antibody combined group
Extended Data Table 3 Probability of superiority among the five cohorts in subgroups
Extended Data Table 4 AEs

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mo, H., Yu, Y., Sun, X. et al. Metronomic chemotherapy plus anti-PD-1 in metastatic breast cancer: a Bayesian adaptive randomized phase 2 trial. Nat Med (2024). https://doi.org/10.1038/s41591-024-03088-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41591-024-03088-2

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer